Skip to main content
Top
Published in: BMC Gastroenterology 1/2020

Open Access 01-12-2020 | Non-Celiac Gluten Sensitivity | Research article

More fuel to the fire: some patients with non-celiac self-reported wheat sensitivity exhibit adaptive immunological responses in duodenal mucosa

Authors: Antonia Isabel Castillo-Rodal, Janette Furuzawa-Carballeda, Mario Peláez-Luna, José Castro-Gómez, Yolanda López-Vidal, Luis Uscanga

Published in: BMC Gastroenterology | Issue 1/2020

Login to get access

Abstract

Background

In contrast to the well-characterized Celiac Disease (CD), the clinical scenarios encompassed by the non-celiac self-reported wheat sensitivity (NCSRWS) might be related to different antigens that trigger distinct immune-inflammatory reactions. Although an increased number of intestinal intraepithelial lymphocytes is observed at the inception of both diseases, the subsequent immunopathogenic pathways seem to be different. We aimed to describe the cytokine profile observed in the duodenal mucosa of patients with NCSRWS.

Methods

In a blind, cross-sectional study, we included duodenal biopsies from 15 consecutive untreated patients with active CD, 9 individuals with NCSRWS and 10 subjects with dyspepsia without CD and food intolerances. Immunohistochemistry and flow-cytometry were used to determine the presence of pro-inflammatory cytokine expressing monocytes and monocyte-derived dendritic cells involved in innate immune activation, cytokine-driven polarization and maintenance of Th1 and Th17/Th 22, and anti-inflammatory/profibrogenic cytokines.

Results

The percentage of cells expressing all tested cytokines in the lamina propria and the epithelium was higher in CD patients than in the control group. Cytokines that induce and maintain Th1 and Th17 polarization were higher in CD than in NCSRWS and controls, also were higher in NCSRWS compared to controls. Similar differences were detected in the expression of IL-4 and TGF-1, while IL-10-expressing cells were lower in NCSRWS patients than in controls and CD subjects.

Conclusions

NCSRWS patients exhibit components of both, innate and adaptive immune mechanisms but to a lesser extent compared to CD.
Literature
1.
go back to reference Gasbarrini GB, Mangiola F. Wheat related disorders:A broadspectrum of evolving diseases. United Eur Gastroenterol J. 2014;2(4):254–62.CrossRef Gasbarrini GB, Mangiola F. Wheat related disorders:A broadspectrum of evolving diseases. United Eur Gastroenterol J. 2014;2(4):254–62.CrossRef
3.
go back to reference Czaja-Bulsa G. Non coeliac gluten sensitivity—a new disease with gluten intolerance. Clin Nutr. 2015;34:189–94.PubMedCrossRef Czaja-Bulsa G. Non coeliac gluten sensitivity—a new disease with gluten intolerance. Clin Nutr. 2015;34:189–94.PubMedCrossRef
5.
go back to reference Carroccio A, Mansueto P, Iacono G, et al. Non-celiac wheat sensitivity diagnosed by double-blind placebo-controlled challenge: exploring a new clinical entity. Am J Gastroenterol. 2012;107:1898–906.PubMedCrossRef Carroccio A, Mansueto P, Iacono G, et al. Non-celiac wheat sensitivity diagnosed by double-blind placebo-controlled challenge: exploring a new clinical entity. Am J Gastroenterol. 2012;107:1898–906.PubMedCrossRef
6.
7.
go back to reference Meresse B, Ripoche J, Heyman M, Cerf-Bensussan N. Celiac disease: from oral tolerance to intestinal inflammation, autoimmunity and lymphomagenesis. Nat Muc Immunol. 2008;2(1):8–23.CrossRef Meresse B, Ripoche J, Heyman M, Cerf-Bensussan N. Celiac disease: from oral tolerance to intestinal inflammation, autoimmunity and lymphomagenesis. Nat Muc Immunol. 2008;2(1):8–23.CrossRef
8.
go back to reference Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel therapies. Gastroenterology. 2009;137:1912–33.PubMedCrossRef Schuppan D, Junker Y, Barisani D. Celiac disease: from pathogenesis to novel therapies. Gastroenterology. 2009;137:1912–33.PubMedCrossRef
9.
go back to reference Kupfer SS, Jabri B. Pathophysiology of celiac disease. Gastrointest Endosc Clin N Am. 2012;22(4):639–60.PubMedCrossRef Kupfer SS, Jabri B. Pathophysiology of celiac disease. Gastrointest Endosc Clin N Am. 2012;22(4):639–60.PubMedCrossRef
10.
11.
go back to reference Stamnaes J, Sollid LM. Celiac disease: autoimmunity in response to food antigen. Semin Immunol. 2015;27:343–52.PubMedCrossRef Stamnaes J, Sollid LM. Celiac disease: autoimmunity in response to food antigen. Semin Immunol. 2015;27:343–52.PubMedCrossRef
12.
go back to reference Caio G, Volta U, Sapone A, et al. Celiac disease: a comprehensive current review. BMC Med. 2019;17:14.CrossRef Caio G, Volta U, Sapone A, et al. Celiac disease: a comprehensive current review. BMC Med. 2019;17:14.CrossRef
13.
go back to reference Nijeboer P, Bontkes HJ, Mulder CJ, Bouma G. Non-celiac gluten sensitivity. Is it the Gluten or the Grain? J Gastrointestin Liver Dis. 2013;22(4):435–40.PubMed Nijeboer P, Bontkes HJ, Mulder CJ, Bouma G. Non-celiac gluten sensitivity. Is it the Gluten or the Grain? J Gastrointestin Liver Dis. 2013;22(4):435–40.PubMed
14.
go back to reference Molina-Infante J, Carroccio A. Suspected non-celiac gluten sensitivity confirmed in few patients after gluten challenge in double-blind, placebo-controlled trials. Clin Gastroenterol Hepatol. 2017;15:339–48.PubMedCrossRef Molina-Infante J, Carroccio A. Suspected non-celiac gluten sensitivity confirmed in few patients after gluten challenge in double-blind, placebo-controlled trials. Clin Gastroenterol Hepatol. 2017;15:339–48.PubMedCrossRef
15.
go back to reference Biesiekierski JR, Peters SL, Newnham ED, Rosella O, Muir J, Gibson PR. No effects of gluten in patients with self-reported non-celiac gluten sensitivity after dietary reduction of fermentable, poor-absorbed short-chain carbohydrates. Gastroenterology. 2013;145:320–8.PubMedCrossRef Biesiekierski JR, Peters SL, Newnham ED, Rosella O, Muir J, Gibson PR. No effects of gluten in patients with self-reported non-celiac gluten sensitivity after dietary reduction of fermentable, poor-absorbed short-chain carbohydrates. Gastroenterology. 2013;145:320–8.PubMedCrossRef
16.
go back to reference Zanini B, Baschè R, Ferraresi A, et al. Randomized clinical study: gluten challenge induces symptom recurrence in only a minority of patients who meet clinical criteria for non-celiac gluten sensitivity. Aliment Pharmacol Ther. 2015;42:968–76.PubMedCrossRef Zanini B, Baschè R, Ferraresi A, et al. Randomized clinical study: gluten challenge induces symptom recurrence in only a minority of patients who meet clinical criteria for non-celiac gluten sensitivity. Aliment Pharmacol Ther. 2015;42:968–76.PubMedCrossRef
17.
go back to reference Skoje GI, Sarna VK, Minelle H, et al. Fructan, rather than gluten, induces symptoms in patients with self-reported non-celiac gluten sensitivity. Gastroenterology. 2018;154:529–39.CrossRef Skoje GI, Sarna VK, Minelle H, et al. Fructan, rather than gluten, induces symptoms in patients with self-reported non-celiac gluten sensitivity. Gastroenterology. 2018;154:529–39.CrossRef
18.
go back to reference Schuppan D, Zevallos V. Wheat amylase trypsin inhibitors as nutritional activators of innate immunity. Dig Dis. 2015;33:260–3.PubMedCrossRef Schuppan D, Zevallos V. Wheat amylase trypsin inhibitors as nutritional activators of innate immunity. Dig Dis. 2015;33:260–3.PubMedCrossRef
19.
go back to reference Mansueto P, Soresi M, Iacobucci R, et al. Non-celiac wheatsensitivity: a search for the pathogenesisof a self-reported condition. Italian J Med. 2019;13:15–23.CrossRef Mansueto P, Soresi M, Iacobucci R, et al. Non-celiac wheatsensitivity: a search for the pathogenesisof a self-reported condition. Italian J Med. 2019;13:15–23.CrossRef
20.
go back to reference Zevallos VF, Raker V, Tenzer S, et al. Nutritional wheat amylase-trypsin inhibitors promote intestinal inflammation via activation of myeloid cells. Gastroenterology. 2017;152:1100–13.PubMedCrossRef Zevallos VF, Raker V, Tenzer S, et al. Nutritional wheat amylase-trypsin inhibitors promote intestinal inflammation via activation of myeloid cells. Gastroenterology. 2017;152:1100–13.PubMedCrossRef
21.
go back to reference Sapone A, Lammer KM, Mazzarella G, et al. Differential mucosal IL-17 expression in two gliadin-induced disorders: gluten sensitivity and the autoimmune enteropathy celiac disease. Int Arch Immunol. 2010;152:75–80.CrossRef Sapone A, Lammer KM, Mazzarella G, et al. Differential mucosal IL-17 expression in two gliadin-induced disorders: gluten sensitivity and the autoimmune enteropathy celiac disease. Int Arch Immunol. 2010;152:75–80.CrossRef
22.
go back to reference Sapone A, Lammers KM, Casolaro V, et al. Divergence of gut permeability and mucosal gene expression in two gluten associated conditions: celiac disease and gluten sensitivity. BMC Med. 2011;9(23):1–11. Sapone A, Lammers KM, Casolaro V, et al. Divergence of gut permeability and mucosal gene expression in two gluten associated conditions: celiac disease and gluten sensitivity. BMC Med. 2011;9(23):1–11.
23.
go back to reference Molina-Infante J, Santolaria S, Montoro M, Esteve M, Fernández-Bañares F. Non-celiac gluten sensitivity: a critical review of current evidence. Gastroenterol Hepatol. 2014;37(6):362–71.PubMedCrossRef Molina-Infante J, Santolaria S, Montoro M, Esteve M, Fernández-Bañares F. Non-celiac gluten sensitivity: a critical review of current evidence. Gastroenterol Hepatol. 2014;37(6):362–71.PubMedCrossRef
24.
go back to reference Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol. 1999;11(1185):1194. Oberhuber G, Granditsch G, Vogelsang H. The histopathology of coeliac disease: time for a standardized report scheme for pathologists. Eur J Gastroenterol Hepatol. 1999;11(1185):1194.
25.
go back to reference Ford AC, Bercik P, Morgan DG, Bolino C, Pintos-Sanchez MI, Moayyedi P. The Rome III criteria for the diagnosis of functional dyspepsia in secondary care are not superior to previous definitions. Gastroenterology. 2014;146(4):932–40.PubMedCrossRef Ford AC, Bercik P, Morgan DG, Bolino C, Pintos-Sanchez MI, Moayyedi P. The Rome III criteria for the diagnosis of functional dyspepsia in secondary care are not superior to previous definitions. Gastroenterology. 2014;146(4):932–40.PubMedCrossRef
26.
go back to reference Valle J, Morgado JMT, Ruíz-Martín J, et al. Flow cytometryofduodenal intraepithelial lymphocytesimproves diagnosis ofceliac diseasein difficultcases. United Eur Gastroenterol J. 2017;5:819–26.CrossRef Valle J, Morgado JMT, Ruíz-Martín J, et al. Flow cytometryofduodenal intraepithelial lymphocytesimproves diagnosis ofceliac diseasein difficultcases. United Eur Gastroenterol J. 2017;5:819–26.CrossRef
27.
go back to reference Furuzawa Carballeda J, Fonseca Camarillo G, Yamamoto-Furusho JK. Interleukin 27 is upregulated in patients with active inflammatory bowel disease. Immunol Res. 2016;64:901–7.PubMedCrossRef Furuzawa Carballeda J, Fonseca Camarillo G, Yamamoto-Furusho JK. Interleukin 27 is upregulated in patients with active inflammatory bowel disease. Immunol Res. 2016;64:901–7.PubMedCrossRef
28.
go back to reference Leon F. Flow cytometry of intestinal intraepithelial lymphocytes in celiac disease. J Immunol Methods. 2011;363(2):177–86.PubMedCrossRef Leon F. Flow cytometry of intestinal intraepithelial lymphocytes in celiac disease. J Immunol Methods. 2011;363(2):177–86.PubMedCrossRef
29.
go back to reference Mansueto P, D’Alcamo A, Seidita A, Carroccio A. Food allergy inirritablebowel syndrome: the case of non-celiacwheat sensitivity. World J Gastroenterol. 2015;21:7089–109.PubMedPubMedCentralCrossRef Mansueto P, D’Alcamo A, Seidita A, Carroccio A. Food allergy inirritablebowel syndrome: the case of non-celiacwheat sensitivity. World J Gastroenterol. 2015;21:7089–109.PubMedPubMedCentralCrossRef
30.
go back to reference Vazquez-Roque MI, Camilleri M, Smyrk T, et al. A controlled trial of gluten free diet in patients with irritable bowel syndrome-diarrhea: effects on bowel frequency and intestinal function. Gastroenterology. 2013;144:903–11.PubMedCrossRef Vazquez-Roque MI, Camilleri M, Smyrk T, et al. A controlled trial of gluten free diet in patients with irritable bowel syndrome-diarrhea: effects on bowel frequency and intestinal function. Gastroenterology. 2013;144:903–11.PubMedCrossRef
31.
go back to reference Figueroa-Salcido OG, Ontiveros N, Cabrera-Chavez F. Glutenvehicle andplacebo for non-celiac sensitivity assessment. Medicina (Kaunas). 2019;55:117.CrossRef Figueroa-Salcido OG, Ontiveros N, Cabrera-Chavez F. Glutenvehicle andplacebo for non-celiac sensitivity assessment. Medicina (Kaunas). 2019;55:117.CrossRef
32.
go back to reference Masaebi F, Azizmohammad Looha M, Rostami-Nejad M, Pourhoseingholi MA, Mohseni N, Samasca G, Lupan I, Rezaei-Tavirani M, Zali MR. The predictive value of serum cytokines for distinguishing celiac disease from non-celiac gluten sensitivity and healthy subjects. Iran Biomed J. 2020;24(4):335–41.CrossRef Masaebi F, Azizmohammad Looha M, Rostami-Nejad M, Pourhoseingholi MA, Mohseni N, Samasca G, Lupan I, Rezaei-Tavirani M, Zali MR. The predictive value of serum cytokines for distinguishing celiac disease from non-celiac gluten sensitivity and healthy subjects. Iran Biomed J. 2020;24(4):335–41.CrossRef
33.
go back to reference Heydari F, Rostami-Nejad M, Moheb-Alian A, Mollahoseini MH, Rostami K, Pourhoseingholi MA, Aghamohammadi E, Zali MR. Serum cytokines profile in treated celiac disease compared with non-celiac gluten sensitivity and control: a marker for differentiation. J Gastrointest Liver Dis. 2018;27(3):241–7.CrossRef Heydari F, Rostami-Nejad M, Moheb-Alian A, Mollahoseini MH, Rostami K, Pourhoseingholi MA, Aghamohammadi E, Zali MR. Serum cytokines profile in treated celiac disease compared with non-celiac gluten sensitivity and control: a marker for differentiation. J Gastrointest Liver Dis. 2018;27(3):241–7.CrossRef
34.
go back to reference Ido H, Matsubara H, Kuroda M, Takahashi A, Kojima Y, Koikeda S, Sasaki M. Combination of gluten-digesting enzymes improved symptoms of non-celiac gluten sensitivity: a randomized single-blind, placebo-controlled crossover study. Clin Transl Gastroenterol. 2018;9(9):181.PubMedPubMedCentralCrossRef Ido H, Matsubara H, Kuroda M, Takahashi A, Kojima Y, Koikeda S, Sasaki M. Combination of gluten-digesting enzymes improved symptoms of non-celiac gluten sensitivity: a randomized single-blind, placebo-controlled crossover study. Clin Transl Gastroenterol. 2018;9(9):181.PubMedPubMedCentralCrossRef
35.
go back to reference Di Sabatino A, Giuffrida P, Fornasa G, Salvatore C, Vanoli A, Naviglio S, De Leo L, Pasini A, De Amici M, Alvisi C, Not T, Rescigno M, Corazza GR. Innate and adaptive immunity in self-reported nonceliac gluten sensitivity versus celiac disease. Dig Liver Dis. 2016;48(7):745–52.PubMedCrossRef Di Sabatino A, Giuffrida P, Fornasa G, Salvatore C, Vanoli A, Naviglio S, De Leo L, Pasini A, De Amici M, Alvisi C, Not T, Rescigno M, Corazza GR. Innate and adaptive immunity in self-reported nonceliac gluten sensitivity versus celiac disease. Dig Liver Dis. 2016;48(7):745–52.PubMedCrossRef
36.
go back to reference Brottveit M, Beitnes AC, Tollefsen S, Bratlie JE, Jahnsen FL, Johansen FE, Sollid LM, Lundin KE. Mucosal cytokine response after short-term gluten challenge in celiac disease and non-celiac gluten sensitivity. Am J Gastroenterol. 2013;108(5):842–50.PubMedCrossRef Brottveit M, Beitnes AC, Tollefsen S, Bratlie JE, Jahnsen FL, Johansen FE, Sollid LM, Lundin KE. Mucosal cytokine response after short-term gluten challenge in celiac disease and non-celiac gluten sensitivity. Am J Gastroenterol. 2013;108(5):842–50.PubMedCrossRef
37.
go back to reference Junker Y, Ziessig S, Kim SJ, et al. Wheatamylase trypsin inhibitorsdrive intestinalinflammation via activationoftoll-likereceptor 4. J Exp Med. 2012;209:2395–408.PubMedPubMedCentralCrossRef Junker Y, Ziessig S, Kim SJ, et al. Wheatamylase trypsin inhibitorsdrive intestinalinflammation via activationoftoll-likereceptor 4. J Exp Med. 2012;209:2395–408.PubMedPubMedCentralCrossRef
38.
go back to reference Zhou W, Zhang F, Aune TM. Either IL-2 or IL-12 is sufficient to direct Th1 differentiation by nonobese Diabetic T Cells. J Immunol. 2003;170:735–40.PubMedCrossRef Zhou W, Zhang F, Aune TM. Either IL-2 or IL-12 is sufficient to direct Th1 differentiation by nonobese Diabetic T Cells. J Immunol. 2003;170:735–40.PubMedCrossRef
39.
go back to reference Feili-Hariri M, Falkner DH, Morel PA. Polarization of naive T cells into Th1 or Th2 by distinct cytokine-driven murine dendritic cell populations: implications for immunotherapy. J Leukoc Biol. 2005;78(3):656–64.PubMedCrossRef Feili-Hariri M, Falkner DH, Morel PA. Polarization of naive T cells into Th1 or Th2 by distinct cytokine-driven murine dendritic cell populations: implications for immunotherapy. J Leukoc Biol. 2005;78(3):656–64.PubMedCrossRef
40.
go back to reference Bosman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012;12:180–90.CrossRef Bosman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol. 2012;12:180–90.CrossRef
41.
go back to reference Mention JJ, Ben Ahmed M, Beque B, et al. Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease. Gastroenterology. 2003;125(3):730–45.PubMedCrossRef Mention JJ, Ben Ahmed M, Beque B, et al. Interleukin 15: a key to disrupted intraepithelial lymphocyte homeostasis and lymphomagenesis in celiac disease. Gastroenterology. 2003;125(3):730–45.PubMedCrossRef
42.
go back to reference Harris KM, Fasano A, Mann DL. Monocytes differentiated with IL-15 support Th17 and Th1 responses to wheat gliadin: implications for celiac disease. Clin Immunol. 2008;135:430–9.CrossRef Harris KM, Fasano A, Mann DL. Monocytes differentiated with IL-15 support Th17 and Th1 responses to wheat gliadin: implications for celiac disease. Clin Immunol. 2008;135:430–9.CrossRef
44.
go back to reference Chong DLW, Ingram RJ, Lowther DE, Muir R, Sriskandan S, Altmann D. The nature of innate and adaptive interleukin-17A responses in sham or bacterial inoculation. Immunology. 2012;136:325–33.PubMedPubMedCentralCrossRef Chong DLW, Ingram RJ, Lowther DE, Muir R, Sriskandan S, Altmann D. The nature of innate and adaptive interleukin-17A responses in sham or bacterial inoculation. Immunology. 2012;136:325–33.PubMedPubMedCentralCrossRef
46.
go back to reference Di Liberto D, Mansueto P, D’Alcamo A. Predominance oftype1 innate lymphoidcells in the rectal mucosa of patients with non-celiacwheat sensitivity: reversal aftera wheat-free diet. Clin Transl Gastroenterol. 2016;7:e178.PubMedPubMedCentralCrossRef Di Liberto D, Mansueto P, D’Alcamo A. Predominance oftype1 innate lymphoidcells in the rectal mucosa of patients with non-celiacwheat sensitivity: reversal aftera wheat-free diet. Clin Transl Gastroenterol. 2016;7:e178.PubMedPubMedCentralCrossRef
47.
go back to reference Volta U, Bardela MT, Calabro A, Troncone R, Corazza GR. Study group for non-celiac gluten sensitivity. An Italian prospective multicenter survey on patients suspected of having non-celiac gluten sensitivity. BMC Med. 2014;12:85.PubMedPubMedCentralCrossRef Volta U, Bardela MT, Calabro A, Troncone R, Corazza GR. Study group for non-celiac gluten sensitivity. An Italian prospective multicenter survey on patients suspected of having non-celiac gluten sensitivity. BMC Med. 2014;12:85.PubMedPubMedCentralCrossRef
48.
go back to reference Schepatti A, Sanders DS, Biagi F. Seronegative coeliac disease: clearing the diagnostic dilemma. Curr Opin Gastroenterol. 2018;34:154–8.CrossRef Schepatti A, Sanders DS, Biagi F. Seronegative coeliac disease: clearing the diagnostic dilemma. Curr Opin Gastroenterol. 2018;34:154–8.CrossRef
Metadata
Title
More fuel to the fire: some patients with non-celiac self-reported wheat sensitivity exhibit adaptive immunological responses in duodenal mucosa
Authors
Antonia Isabel Castillo-Rodal
Janette Furuzawa-Carballeda
Mario Peláez-Luna
José Castro-Gómez
Yolanda López-Vidal
Luis Uscanga
Publication date
01-12-2020
Publisher
BioMed Central
Published in
BMC Gastroenterology / Issue 1/2020
Electronic ISSN: 1471-230X
DOI
https://doi.org/10.1186/s12876-020-01564-w

Other articles of this Issue 1/2020

BMC Gastroenterology 1/2020 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.